Discovery and Preclinical Evaluation of BMS ... - ACS Publications

Apr 4, 2016 - Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, New Jersey 08540, United States. •S Supporting ...
0 downloads 0 Views 1019KB Size
Subscriber access provided by UOW Library

Letter

Discovery and Preclinical Evaluation of BMS-711939, an Oxybenzyl-glycine Based PPAR# Selective Agonist Yan Shi, Jun Li, Lawrence J Kennedy, Shiwei Tao, Andres S. Hernandez, Zhi Lai, Sean Chen, Henry Wong, Juliang Zhu, Ashok Trehan, Ngiap-Kie Lim, Huiping Zhang, Bang-Chi Chen, Kenneth T Locke, Kevin M O'Malley, Litao Zhang, Raiajit Srivastava, Bowman Miao, Daniel S. Meyers, Hossain Monshizadegan, Debra Search, Denise Grimm, Rongan Zhang, Thomas Harrity, Lori K Kunselman, Michael Cap, Jodi Muckelbauer, Chiehying Chang, Stanley R. Krystek, YiXin Li, Vinayak Hosagrahara, Lisa Zhang, Pathanjali Kadiyala, Carrie Xu, Michael A Blanar, Robert Zahler, Ranjan Mukherjee, Peter T.W. Cheng, and Joseph A Tino ACS Med. Chem. Lett., Just Accepted Manuscript • DOI: 10.1021/acsmedchemlett.6b00033 • Publication Date (Web): 04 Apr 2016 Downloaded from http://pubs.acs.org on April 7, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Medicinal Chemistry Letters is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 12

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Medicinal Chemistry Letters

Zhang, Rongan; Bristol-Myers Squibb Pharmaceutical Research and Development Harrity, Thomas; Bristol-Myers Squibb Pharmaceutical Research and Development Kunselman, Lori; Bristol-Myers Squibb Pharmaceutical Research and Development Cap, Michael; Bristol-Myers Squibb Pharmaceutical Research and Development Muckelbauer, Jodi; Bristol-Myers Squibb Pharmaceutical Research and Development Chang, Chiehying; Bristol-Myers Squibb Pharmaceutical Research and Development Krystek, Stanley; Bristol-Myers Squibb, Computer-Assisted Drug Design Li, YiXin; Bristol-Myers Squibb Pharmaceutical Research and Development Hosagrahara, Vinayak; Bristol-Myers Squibb Pharmaceutical Research and Development Zhang, Lisa; Bristol-Myers Squibb Pharmaceutical Research and Development Kadiyala, Pathanjali; Bristol-Myers Squibb Pharmaceutical Research and Development Xu, Carrie; Bristol-Myers Squibb Pharmaceutical Research and Development Blanar, Michael; Bristol-Myers Squibb Pharmaceutical Research and Development Zahler, Robert; Bristol-Myers Squibb Pharmaceutical Research and Development Mukherjee, Ranjan; Bristol-Myers Squibb Pharmaceutical Research and Development Cheng, Peter; Bristol-Myers Squibb Pharmaceutical Research Institute, Metabolic Diseases Chemistry Tino, Joseph; Bristol-Myers Squibb Pharmaceutical Research and Development

ACS Paragon Plus Environment

ACS Medicinal Chemistry Letters

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 12

Discovery and Preclinical Evaluation of BMS-711939, an Oxybenzylglycine Based PPARα Selective Agonist Yan Shi,* Jun Li, Lawrence J. Kennedy, Shiwei Tao, Andrés S. Hernández, Zhi Lai, Sean Chen, Henry Wong, Juliang Zhu, Ashok Trehan, Ngiap-Kie Lim, Huiping Zhang, Bang-Chi Chen, Kenneth T. Locke, Kevin M. O'Malley, Litao Zhang, Raiajit Srivastava, Bowman Miao, Daniel S. Meyers, Hossain Monshizadegan, Debra Search, Denise Grimm, Rongan Zhang, Thomas Harrity, Lori K. Kunselman, Michael Cap, Jodi Muckelbauer, Chiehying Chang, Stanley R. Krystek, Yi-Xin Li, Vinayak Hosagrahara, Lisa Zhang, Pathanjali Kadiyala, Carrie Xu, Michael A. Blanar, Robert Zahler, Ranjan Mukherjee, Peter T. W. Cheng, Joseph A. Tino Research and Development, Bristol-Myers Squibb Company, 350 Carter Road, Hopewell, NJ 08540, U.S.A. KEYWORDS: peroxisome proliferator-activated receptor (PPAR) α selective agonist, high fat fed hamster model, human ApoA1 transgenic mice, pharmacokinetics. ABSTRACT: BMS-711939 (3) is a potent and selective peroxisome proliferator-activated receptor (PPAR) α agonist, with an EC50 of 4 nM for human PPARα and >1000-fold selectivity vs. human PPARγ (EC50 = 4.5 µM) and PPARδ (EC50 > 100 µM) in PPAR-GAL4 transactivation assays. Compound 3 also demonstrated excellent in vivo efficacy and safety profiles in preclinical studies and thus was chosen for further preclinical evaluation. The synthesis, structure–activity relationship (SAR) studies, and in vivo pharmacology of 3 in preclinical animal models as well as its ADME profile are described.

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors in the nuclear hormone receptor superfamily.1, 2 Three subtypes of PPARs, namely PPARα, PPARγ and PPARδ, have been identified in various species, including humans. PPARα is highly expressed in the liver and regulates the expression of genes encoding lipid and lipoprotein metabolism.3 Upon binding of PPARα ligand agonists, the resulting conformational change leads to the modulation of a number of PPARα responsive genes, which in turn have pleiotropic effects on plasma lipoprotein levels, atherosclerosis and inflammation. There are currently several synthetic PPARα ligands, including the fibrate class of hypolipidemic drugs in clinical use.4, 5 Although fibrates are ligands of PPARs, their binding affinity to PPARα is relatively weak.6, 7 This results in the relatively high doses (e.g. 200 mg of fenofibrate and 1.2 g of gemfibrozil) needed to achieve clinical efficacy, and unwanted side effects may occur at these high doses. Several potent PPARα selective agonists have been progressed into various phases of clinical development, including GW5907358 and LY518674.9 However, the development of most of these potent and selective PPARα agonist clinical candidates have been suspended. Recently, the PPARα-selective α,γ dual agonist saroglitazar was approved in India for the treatment of diabetic dyslipidemia.10 A potent and efficacious PPARα agonist with an excellent safety profile may pro-

vide an opportunity for the treatment of atherosclerosis and dyslipidemia as well as non-alcoholic steatohepatitis (NASH)11 with minimized side effects. In addition, we considered that a more potent and selective PPARα agonist with minimized disparity in interspecies PPAR activities (particularly rodent), will be useful for mechanismbased safety evaluation in preclinical animal models. Figure 1. Fenofibric acid and BMS-687453

In continuation of our research in the field of PPARs to develop novel therapeutic agents to treat metabolic disorders,12 we have reported the discovery of the selective PPARα agonist 213 (Figure 1), which exhibited a high degree of selectivity towards PPARα over PPARγ (PPARα EC50 = 10 nM, EC50 ratio: γ/α = 410), and demonstrated an excellent pharmacological and safety profile in preclinical studies. Inspection of the X-ray crystal structures of 2 bound to the PPARα ligand binding domain (LBD; Figure 2) suggested that the carboxylic acid of 2 forms the wellrecognized hydrogen-bonding network with neighboring residues: His440, Tyr464, Tyr314 and Ser280. The 1,3oxybenzyl central core anchors the acid head group and

ACS Paragon Plus Environment

Page 3 of 12

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Medicinal Chemistry Letters

the 4-chlorophenyl oxazole moiety into their appropriate binding pockets. Since the central phenyl core and the benzylic carbon are partially surrounded by hydrophobic residues such as Leu321, Ile317 and Phe318, we envioned that small hydrophobic substituents on this region may be tolerated and SAR investigations could be productively conducted within this portion of the molecule to further optimize the binding affinity/potency and/or reduce the human/rodent species difference in PPARα activity. In this paper, we describe the SAR findings from this effort, which resulted in the discovery of 3 (BMS-711939). We also disclose the pharmacokinetic and in vivo efficacy profiling of 3, which progressed into preclinical toxicology studies.

action of 23a-c with methyl chloroformate gave the corresponding methyl carbamates 24a-c in 90-99% yield. Basemediated alkylation of phenols 24a-c with 4(chloromethyl)-2-(4-chlorophenyl)-5-methyloxazole 25a at 80oC provided the methyl esters 26a-c in 86-99% yield. Basic hydrolysis of 26a-c provided 3-5 in 90-93% yield. Compound 6 was synthesized in a similar fashion from intermediate 27.14 Compounds 7-13 were synthesized from intermediate 24a and oxazoles 25b-h15 using the same synthetic sequence as described for 3-5. Compounds 14-20 were synthesized through a slightly different sequence as shown in Scheme 2. Alkylation of 2fluoro-5-hydroxybenzaldehyde 21a with 4-(chloromethyl) oxazole 25a provided aldehyde 28 in 95% yield. Reductive amination of 28 with 22 afforded the secondary amine 29 in 90% yield. Reaction of 29 with different chloroformates and subsequent hydrolysis of the resulting esters provided compounds 14-20 in 80-93% yield. O HO

O H

25a

O Cl N

Me

O HO

a

H H2N

2

4

F 3 21a, 2-F 21b, 3-F 21c, 4-F

22

N F

O

CO2Me

23a, 2-F 23b, 3-F 23c, 4-F Me

Cl

c

O

N

O

O 1

O 5

R N

F 3

O

CO2H O

O

27

CO2Me

c, d, e

Cl N

Cl

N 25a, 25b, 25c, 25d,

3, 2-F 4, 3-F 5, 4-F

N

CO2Me O

Me

N

O

O

26a, 2-F F 26b, 3-F 26c, 4-F

Cl

HO

N

O

24a, 2-F 24b, 3-F 24c, 4-F d, e

b

CO2Me

F

O

HO

N H

CO2Me

R = Cl R = Me R= F R= H

25e, 25f, 25g, 25h,

R = CF3 R = OCF3 R = t-Bu R = SO2Me

Me O

O

N O

CO2H O

6

Scheme 1. The synthesis of 3-6. Reagents and conditions: (a) Et3N, MeOH, NaBH4, 0°C to rt. (b) methyl chloroformate, aq. NaHCO3, THF. (c) 25a, K2CO3, MeCN, 80°C. (d) LiOH, THF-H2O, rt. (e) 1N aq. HCl.

The syntheses of 3-6 are described in Scheme 1. Reductive amination of fluoro-substituted hydroxybenzaldehydes 21a-c with glycine methyl ester hydrochloride 22 afforded the secondary amines 23a-c in 85-95% yield. Re-

Me

O O

H

28

HN O

29

HO

N

F

21a

Figure 2. X-ray crystal structure of 2 bound in PPARα LBD with 2.7Å resolution. Residues within 4.5 Å of benzylglycine are shown with thick sticks. Hydrogen bonds are shown as black dashed lines. The PDB deposition number is 3KDT.

Cl

Cl

N

F

b

a

O + Cl

O

CO2Me c, d, e

O Cl N

F

14: 15: 16: 17: 18: 19: 20:

Me

R

O

CO2H N

O

R = Et R = n-Pr R = F(CH2)3 R = n-Bu R = i-Bu R = 4-MePh R = 4-MeOPh

F

Scheme 2. The synthesis of 14-20. Reagents and conditions: (a) K2CO3, MeCN, 80°C. (b) 22, Et3N, MeOH, NaBH4, 0°C to rt. (c) ROCOCl, aq. NaHCO3, THF. (d) LiOH, THF-H2O, rt. (e) 1N aq. HCl.

Our initial effort to further optimize 2 focused on structural modifications of the central core and the benzylic position of the benzylglycine. The SAR results are shown in Table 1. Introducing a fluorine atom at the 2-position of central core resulted in a very potent and selective PPARα/γ agonist 3 (EC50 γ/α ratio = 1128), with PPARα EC50 = 4 nM and PPARγ EC50 = 4.51 µM in the transactivation assays.13 This compound is ~2.5-fold more potent than 2 (EC50 = 10 nM) at PPARα, and is slightly less potent at PPARγ than 2 (PPARγ EC50 = 4.1 µM). These functional data correspond well to their PPARα and PPARγ binding affinities (Table 1).16 On the other hand, the opposite effects were observed when introducing a fluorine atom at the 3-position of central phenyl core; compound 4 (EC50 γ/α ratio = 157) is less potent at PPARα (EC50 = 18 nM) and more potent at PPARγ (EC50 = 2.82 µM) than 2. Interestingly, the 4-fluoro-substituted analog 5 is less active than 2 at both PPARα and PPARγ with a γ/α EC50 ratio of 98. When a (S)-methyl group is placed at the α-position of the benzylglycine of 2, the resulting 6 (EC50 γ/α ratio = 332) has similar potency at PPARα but is more potent at PPARγ than 2. These results indicate that a subtle substitution change on the central phenyl core or at the benzylic position can result in changes in both the PPARα and PPARγ agonist activities and thus the PPARγ/α selec-

ACS Paragon Plus Environment

ACS Medicinal Chemistry Letters

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

tivety. The 2-fluorophenyl glycine analog 3 has the most potent PPARα agonist activity and is also the most selective PPARα agonist among the three isomeric monofluorinated analogs.

Page 4 of 12

ingly, a bulky aliphatic substituent such as t-butyl (12) significantly improved PPARγ potency while maintaining PPARα activity, thus resulting in a significant reduction in PPARα selectivity versus 3. These results suggested that polarity and steric effects play important roles in determining PPARα/γ functional activities and selectivity in this portion of the molecule. This SAR study of the “lefthand” phenyl ring substituents showed that the 4-Clphenyl moiety was optimal, providing potent PPARα activity and the highest level of PPARα selectivity. The corresponding SAR study of the “right-hand” side Cp d

Table 1. Transactivation EC50 and binding IC50 of 2-6a

Cp d 2

1

R

R

αEC50 (nM)

2

γEC50 (nM)

γ/α Ratio

3

4-Cl

Me

4.0

4511

1128

7

4-Me

Me

2.8

2182

787

8

4-F

Me

14.5

10010

691

9

4-H

Me

59

8607

146

10

4-CF3

Me

3.6

2062

566

11

4-CF3O

Me

4.3

2384

556

R

αEC50 (nM)

γEC50 (nM)

γ/α EC5 0 ratio

αIC50 (nM)

γIC50 (nM)

12

4C(CH3)3

Me

2.7

414

154

>2500

>25000

NA

4096

410

260

4-MeSO2

Me

10

>4800 0

13

H

14

4-Cl

Et

4.0

4273

1085

15

4-Cl

n-Pr

3.4

2013

594

16

4-Cl

F(CH2)3

10.6

4383

413

17

4-Cl

n-Bu

2.8

1235

435

18

4-Cl

i-Bu

2.3

1504

666

19

4-Cl

4-MePh

5.6

1255

224

20

4-Cl

4-MeOPh

6.8

1087

160

3

2-F

4.0

4511

1128

97

>9840 0

4

3-F

18

2819

157

1341

>15000

5

4-F

94

9168

98

1563

>15000

6

-

8.6

2857

332

671

>15000

a

Compounds were tested for agonist activity on hPPARGAL4 HEK transactivation assay. Transactivation efficacy was defined as percentage of maximum activity as compared with an appropriate standard set at 100% (100 µM fenofibric acid for PPARα, 1.0 µM rosiglitazone for PPARγ). Full PPARα intrinsic activity (>50% relative to 100 µM fenofibric acid) was observed for all tested compounds (n = 1-3).

With lead compound 3 in hand, we systematically investigated the SAR of the “left-hand” phenyl ring substituents and the “right-hand” carbamate substituents, as shown in Table 2. In general, all the 2-fluoro-phenyl central core based compounds show enhanced PPARα agonist activity and PPARα/γ selectivity versus their corresponding unsubstituted phenyl analogs.13 On the “lefthand” phenyl ring (of the oxazole), while the compound without a para substituent (9, PPARα EC50= 59 nM, γ/α ratio = 146) is ~15-fold less potent and far less selective than 3, the para-fluoro compound 8 is only about 3.5-fold less potent than 3 at PPARα and maintains high selectivity (PPARα EC50= 15 nM, γ/α ratio = 691). Compounds with a non-polar substituent (7, 10-12) at the para position generally have similar PPARα potencies to 3. On the other hand, the relatively bulky polar methylsulfonyl substituent (13) obliterates both PPARα and γ activities. Interest-

carbamate showed that simple alkyl carbamates (14-20) generally provide excellent PPARα potency and selectivity (EC50 γ/α ratio > 400-fold), with the ethyl carbamate 14 having almost the same PPARα potency and selectivity (PPARα EC50= 4 nM, γ/α ratio = 1085) as 3. Interestingly, replacing the terminal methyl group of the n-propyl carbamate 15 with a fluoromethyl (16) resulted in a >2-fold decrease in potency at both PPARα and γ, but retained the high γ/α selectivity ratio. The aryl carbamates (19 & 20) were also potent PPARα agonists, but with slightly lower γ/α selectivity compared to the simple alkyl carbamates. The overall SAR in this carbamate region is similar to that previously observed with 2, with the methyl carbamate being optimal.13 Table 2. In vitro activity of 2-fluoro substituted benzylglycine 3, 7-20a

ACS Paragon Plus Environment

Page 5 of 12

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

a

ACS Medicinal Chemistry Letters

See notes for Table 1.

The PPARα potency and selectivity of 3 were further confirmed by testing it in co-transfection assays in HepG2 cells using full length human PPARα and PPARγ receptors.13 In these assays, compound 3 showed excellent PPARα potency (EC50 = 5 nM) with > 320-fold selectivity vs. PPARγ (EC50 > 1.61 µM). These results correlated well with the data observed from the primary GAL-4 HEK transactivation based screening assays as shown in Table 1. Compound 3 also is a full PPARα agonist in the rodent assays, and is > 2.5-fold more potent than 2 in rodent PPARα functional assays, with EC50s of 178 nM, 144 nM and 123 nM for hamster, mouse and rat, respectively.17 Importantly, compound 3 showed negligible activity (EC50 for transactivation >25 µM and efficacies